Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Microbiol Spectr ; 11(6): e0302623, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37982636

ABSTRACT

IMPORTANCE: Astroviruses (AstV) are known suspects of enteric disease in humans and livestock. Recently, AstV have been linked to encephalitis in immunocompromised patients and other animals, such as cattle, minks, and swine. In our study, we also identified AstV in the respiratory samples of calves with signs of bronchopneumonia, suggesting that their tropism could be even broader. We obtained one bovine AstV (BAstV) complete genome sequence by next-generation sequencing and showed that respiratory and enteric AstV from different species formed a divergent genetic cluster with AstV isolated from encephalitis cases, indicating that tropism might be strain-specific. These data provide further insight into understanding the biology of these understudied pathogens and suggest BAstV as a potential new candidate for bovine respiratory disease.


Subject(s)
Astroviridae Infections , Astroviridae , Bronchopneumonia , Cattle Diseases , Encephalitis , Animals , Cattle , Humans , Swine , Astroviridae Infections/veterinary , Bronchopneumonia/veterinary , Virome , Phylogeny , Astroviridae/genetics , Cattle Diseases/diagnosis , Respiratory System , Feces
2.
Infect Genet Evol ; 113: 105483, 2023 09.
Article in English | MEDLINE | ID: mdl-37482235

ABSTRACT

Bovine Parainfluenza Type 3 virus (BPIV-3) is an enveloped, non-segmented single-stranded, negative-sense RNA virus belonging to the Paramyxoviridae family (genus Respirovirus) with a well-known role in Bovine Respiratory Disease (BRD) onset. Being isolated for the first time in 1959, BPIV-3 currently circulates worldwide in cattle herds and is routinely tested in suspected BRD cases. Different commercial vaccines are available to prevent infection and/or to reduce the clinical signs associated with BPIV-3 infection, which are essential to prevent secondary infections. Despite years of molecular surveillance, a very limited number of complete genome sequences were made publicly available, preventing thus the understanding of the genetic diversity of the circulating strains in the field. In addition, no data about the genetic identity between field and vaccine strains is currently available. In this study, we sequenced the full-genome and genetically characterized BPIV-3 strains isolated from animals displaying respiratory illness in France and Sweden, as well as the vaccine strains contained in three different commercialized vaccines. Our results show that the sequences from France and Sweden belong to genotype C. However, a third sequence from Sweden from 2017 clustered within genotype A. The sequencing of vaccine strains revealed that two of the vaccine strains clustered within genotype C, whereas the third vaccine strain belonged to genotype A. Altogether, our findings suggest that both genotypes A and C circulate in Europe and that BPIV-3 field and vaccine strains are genetically divergent. Our sequencing results could be useful to better understand the genetic differences between the circulating field and vaccine BPIV-3 strains. This is crucial for a correct interpretation of diagnostic findings and for the assessment of BPIV-3 prevalence in cattle population.


Subject(s)
Cattle Diseases , Paramyxoviridae Infections , Viral Vaccines , Cattle , Animals , Respirovirus/genetics , Parainfluenza Virus 3, Bovine/genetics , Viral Vaccines/genetics , Europe , Cattle Diseases/epidemiology , Cattle Diseases/prevention & control
3.
J Virol ; 97(2): e0142322, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36692289

ABSTRACT

Concurrent infections with multiple pathogens are often described in cattle with respiratory illness. However, how the host-pathogen interactions influence the clinical outcome has been only partially explored in this species. Influenza D virus (IDV) was discovered in 2011. Since then, IDV has been detected worldwide in different hosts. A significant association between IDV and bacterial pathogens in sick cattle was shown in epidemiological studies, especially with Mycoplasma bovis. In an experimental challenge, IDV aggravated M. bovis-induced pneumonia. However, the mechanisms through which IDV drives an increased susceptibility to bacterial superinfections remain unknown. Here, we used the organotypic lung model precision-cut lung slices to study the interplay between IDV and M. bovis coinfection. Our results show that a primary IDV infection promotes M. bovis superinfection by increasing the bacterial replication and the ultrastructural damages in lung pneumocytes. In our model, IDV impaired the innate immune response triggered by M. bovis by decreasing the expression of several proinflammatory cytokines and chemokines that are important for immune cell recruitment and the bacterial clearance. Stimulations with agonists of cytosolic helicases and Toll-like receptors (TLRs) revealed that a primary activation of RIG-I/MDA5 desensitizes the TLR2 activation, similar to what was observed with IDV infection. The cross talk between these two pattern recognition receptors leads to a nonadditive response, which alters the TLR2-mediated cascade that controls the bacterial infection. These results highlight innate immune mechanisms that were not described for cattle so far and improve our understanding of the bovine host-microbe interactions and IDV pathogenesis. IMPORTANCE Since the spread of the respiratory influenza D virus (IDV) infection to the cattle population, the question about the impact of this virus on bovine respiratory disease (BRD) remains still unanswered. Animals affected by BRD are often coinfected with multiple pathogens, especially viruses and bacteria. In particular, viruses are suspected to enhance secondary bacterial superinfections. Here, we use an ex vivo model of lung tissue to study the effects of IDV infection on bacterial superinfections. Our results show that IDV increases the susceptibility to the respiratory pathogen Mycoplasma bovis. In particular, IDV seems to activate immune pathways that inhibit the innate immune response against the bacteria. This may allow M. bovis to increase its proliferation and to delay its clearance from lung tissue. These results suggest that IDV could have a negative impact on the respiratory pathology of cattle.


Subject(s)
Cattle Diseases , Host Microbial Interactions , Mycoplasma Infections , Orthomyxoviridae Infections , Signal Transduction , Thogotovirus , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/virology , Lung/immunology , Lung/microbiology , Lung/virology , Mycoplasma bovis/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Signal Transduction/immunology , Superinfection/immunology , Superinfection/veterinary , Toll-Like Receptor 2 , Host Microbial Interactions/immunology , Mycoplasma Infections/immunology , Mycoplasma Infections/virology
4.
Virus Evol ; 8(2): veac081, 2022.
Article in English | MEDLINE | ID: mdl-36533151

ABSTRACT

Influenza D virus (IDV) is an emerging influenza virus that was isolated for the first time in 2011 in the USA from swine with respiratory illness. Since then, IDV has been detected worldwide in different animal species, and it was also reported in humans. Molecular epidemiological studies revealed the circulation of two major clades, named D/OK and D/660. Additional divergent clades have been described but have been limited to specific geographic areas (i.e. Japan and California). In Europe, IDV was detected for the first time in France in 2012 and subsequently also in Italy, Luxembourg, Ireland, the UK, Switzerland, and Denmark. To understand the time of introduction and the evolutionary dynamics of IDV on the continent, molecular screening of bovine and swine clinical samples was carried out in different European countries, and phylogenetic analyses were performed on all available and newly generated sequences. Until recently, D/OK was the only clade detected in this area. Starting from 2019, an increase in D/660 clade detections was observed, accompanied by an increase in the overall viral genetic diversity and genetic reassortments. The time to the most recent common ancestor (tMRCA) of all existing IDV sequences was estimated as 1995-16 years before its discovery, indicating that the virus could have started its global spread in this time frame. Despite the D/OK and D/660 clades having a similar mean tMRCA (2007), the mean tMRCA for European D/OK sequences was estimated as January 2013 compared to July 2014 for European D/660 sequences. This indicated that the two clades were likely introduced on the European continent at different time points, as confirmed by virological screening findings. The mean nucleotide substitution rate of the hemagglutinin-esterase-fusion (HEF) glycoprotein segment was estimated as 1.403 × 10-3 substitutions/site/year, which is significantly higher than the one of the HEF of human influenza C virus (P < 0.0001). IDV genetic drift, the introduction of new clades on the continent, and multiple reassortment patterns shape the increasing viral diversity observed in the last years. Its elevated substitution rate, diffusion in various animal species, and the growing evidence pointing towards zoonotic potential justify continuous surveillance of this emerging influenza virus.

5.
Vet Res ; 53(1): 70, 2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36068558

ABSTRACT

Bovine respiratory disease (BRD) is one of the most important diseases impacting the global cattle industry, resulting in significant economic loss. Commonly referred to as shipping fever, BRD is especially concerning for young calves during transport when they are most susceptible to developing disease. Despite years of extensive study, managing BRD remains challenging as its aetiology involves complex interactions between pathogens, environmental and host factors. While at the beginning of the twentieth century, scientists believed that BRD was only caused by bacterial infections ("bovine pasteurellosis"), we now know that viruses play a key role in BRD induction. Mixtures of pathogenic bacteria and viruses are frequently isolated from respiratory secretions of animals with respiratory illness. The increased diagnostic screening data has changed our understanding of pathogens contributing to BRD development. In this review, we aim to comprehensively examine experimental evidence from all existing studies performed to understand coinfections between respiratory pathogens in cattle. Despite the fact that pneumonia has not always been successfully reproduced by in vivo calf modelling, several studies attempted to investigate the clinical significance of interactions between different pathogens. The most studied model of pneumonia induction has been reproduced by a primary viral infection followed by a secondary bacterial superinfection, with strong evidence suggesting this could potentially be one of the most common scenarios during BRD onset. Different in vitro studies indicated that viral priming may increase bacterial adherence and colonization of the respiratory tract, suggesting a possible mechanism underpinning bronchopneumonia onset in cattle. In addition, a few in vivo studies on viral coinfections and bacterial coinfections demonstrated that a primary viral infection could also increase the pathogenicity of a secondary viral infection and, similarly, dual infections with two bacterial pathogens could increase the severity of BRD lesions. Therefore, different scenarios of pathogen dynamics could be hypothesized for BRD onset which are not limited to a primary viral infection followed by a secondary bacterial superinfection.


Subject(s)
Bovine Respiratory Disease Complex , Cattle Diseases , Coinfection , Pasteurella Infections , Respiratory Tract Diseases , Superinfection , Virus Diseases , Animals , Bacteria , Cattle , Cattle Diseases/microbiology , Coinfection/veterinary , Pasteurella Infections/veterinary , Respiratory System , Respiratory Tract Diseases/veterinary , Superinfection/veterinary , Virus Diseases/veterinary
6.
Transbound Emerg Dis ; 69(3): 1227-1245, 2022 May.
Article in English | MEDLINE | ID: mdl-33764631

ABSTRACT

BACKGROUND: Influenza D virus (IDV), a segmented single-stranded negative-sense ribonucleic acid (RNA) virus, belongs to the new Delta influenza virus genus of the Orthomyxoviridae family. Cattle were proposed as the natural reservoir of IDV in which infection was associated with mild-to-moderate respiratory clinical signs (i.e. cough, nasal discharge and dyspnoea). METHODS AND PRINCIPAL FINDINGS: In order to investigate the role of IDV in bovine respiratory disease, during the period 2017-2020, 883 nasal or naso-pharyngeal swabs from Canadian cattle with respiratory signs (cough and/or dyspnoea) were tested by (RT-)qPCR for IDV and other major bovine viral (bovine herpesvirus 1, bovine viral diarrhoea virus, bovine respiratory syncytial virus, bovine parainfluenza virus 3 and bovine coronavirus) and bacterial (Mannheimia haemolytica, Pasteurella multocida, Histophilus somni and Mycoplasma bovis) respiratory pathogens. In addition, whole-genome sequencing and phylogenetic analyses were carried out on five IDV-positive samples. The prevalence of IDV RT-qPCR (with cut-off: Cq < 38) at animal level was estimated at 5.32% (95% confidence interval: 3.94-7.02). Positive result of IDV was significantly associated with (RT-)qPCR-positive results for bovine respiratory syncytial virus and Mycoplasma bovis. While phylogenetic analyses indicate that most segments belonged to clade D/660, reassortment between clades D/660 and D/OK were evidenced in four samples collected in 2018-2020. CONCLUSIONS AND SIGNIFICANCE: Relative importance of influenza D virus and associated pathogens in bovine respiratory disease of Canadian dairy cattle was established. Whole-genome sequencing demonstrated evidence of reassortment between clades D/660 and D/OK. Both these new pieces of information claim for more surveillance of IDV in cattle production worldwide.


Subject(s)
Cattle Diseases/virology , Orthomyxoviridae Infections/veterinary , Respiratory Tract Diseases/veterinary , Thogotovirus/genetics , Animals , Cattle , Cattle Diseases/epidemiology , Cough/veterinary , Disease Reservoirs , Dyspnea/veterinary , Nasal Mucosa/virology , Nasopharynx/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Quebec/epidemiology , Reassortant Viruses/genetics , Respiratory Tract Diseases/epidemiology , Respiratory Tract Diseases/virology , Thogotovirus/classification
7.
Microbiol Spectr ; 9(3): e0169021, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34937196

ABSTRACT

Bovine respiratory disease (BRD) is a major disease of young cattle whose etiology lies in complex interactions between pathogens and environmental and host factors. Despite a high frequency of codetection of respiratory pathogens in BRD, data on the molecular mechanisms and pathogenesis associated with viral and bacterial interactions are still limited. In this study, we investigated the effects of a coinfection with influenza D virus (IDV) and Mycoplasma bovis in cattle. Naive calves were infected by aerosol with a French IDV strain and an M. bovis strain. The combined infection shortened the incubation period, worsened the disease, and led to more severe macroscopic and microscopic lesions compared to these parameters in calves infected with only one pathogen. In addition, IDV promoted colonization of the lower respiratory tract (LRT) by M. bovis and increased white cell recruitment to the airway lumen. The transcriptomic analysis highlighted an upregulation of immune genes in the lungs of coinfected calves. The gamma interferon (IFN-γ) gene was shown to be the gene most statistically overexpressed after coinfection at 2 days postinfection (dpi) and at least until 7 dpi, which correlated with the high level of lymphocytes in the LRT. Downregulation of the PACE4 and TMPRSS2 endoprotease genes was also highlighted, being a possible reason for the faster clearance of IDV in the lungs of coinfected animals. Taken together, our coinfection model with two respiratory pathogens that when present alone induce moderate clinical signs of disease was shown to increase the severity of the disease in young cattle and a strong transcriptomic innate immune response in the LRT, especially for IFN-γ. IMPORTANCE Bovine respiratory disease (BRD) is among the most prevalent diseases in young cattle. BRD is due to complex interactions between viruses and/or bacteria, most of which have a moderate individual pathogenicity. In this study, we showed that coinfection with influenza D virus (IDV) and Mycoplasma bovis increased the severity of the respiratory disease in calves in comparison with IDV or M. bovis infection. IDV promoted M. bovis colonization of the lower respiratory tract and increased white cell recruitment to the airway lumen. The transcriptomic analysis highlighted an upregulation of immune genes in the lungs of coinfected calves. The IFN-γ gene in particular was highly overexpressed after coinfection, correlated with the disease severity, immune response, and white cell recruitment in the lungs. In conclusion, we showed that IDV facilitates coinfections within the BRD complex by modulating the local innate immune response, providing new insights into the mechanisms involved in severe respiratory diseases.


Subject(s)
Bovine Respiratory Disease Complex/pathology , Coinfection/pathology , Immunity, Innate/immunology , Mycoplasma Infections/veterinary , Orthomyxoviridae Infections/veterinary , Animals , Bovine Respiratory Disease Complex/microbiology , Cattle , Coinfection/immunology , Coinfection/microbiology , Interferon-gamma/immunology , Mycoplasma Infections/pathology , Mycoplasma bovis/immunology , Orthomyxoviridae Infections/pathology , Severity of Illness Index , Thogotovirus/immunology
8.
Microorganisms ; 9(12)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34946167

ABSTRACT

Bats are natural hosts for numerous zoonotic viruses, including henipaviruses, which are highly pathogenic for humans, livestock, and other mammals but do not induce clinical disease in bats. Pteropus bats are identified as a reservoir of henipaviruses and the source of transmission of the infection to humans over the past 20 years. A better understanding of the molecular and cellular mechanisms allowing bats to control viral infections requires the development of relevant, stable, and permissive cellular experimental models. By applying a somatic reprogramming protocol to Pteropus bat primary cells, using a combination of ESRRB (Estrogen Related Receptor Beta), CDX2 (Caudal type Homeobox 2), and c-MYC (MYC proto-oncogene) transcription factors, we generated bat reprogrammed cells. These cells exhibit stem cell-like characteristics and neural stem cell molecular signature. In contrast to primary fibroblastic cells, these reprogrammed stem cells are highly permissive to henipaviruses and exhibit specific transcriptomic profiles with the particular expression of certain susceptibility factors such as interferon-stimulated genes (ISG), which may be related to viral infection. These Pteropus bat reprogrammed stem cells should represent an important experimental tool to decipher interactions during henipaviruses infection in Pteropus bats, facilitate isolation and production of bat-borne viruses, and to better understand the bat biology.

9.
Transbound Emerg Dis ; 68(3): 1125-1135, 2021 May.
Article in English | MEDLINE | ID: mdl-32871031

ABSTRACT

Influenza D virus (IDV) is a novel orthomyxovirus that was first isolated in 2011 in the United States from a swine exhibiting influenza-like disease. To date, its detection is extended to all continents and in a broad host range: IDV is circulating in cattle, swine, feral swine, camelids, small ruminants and horses. Evidence also suggests a possible species jump to humans, underlining the issue of zoonotic potential. In Europe, serological investigations in cattle have partially allowed the understanding of the virus diffusion in different countries such as Italy, France, Luxembourg and Ireland. The infection is widespread in cattle but limited in other investigated species, consolidating the assumption of cattle as IDV primary host. We hypothesize that commercial livestock trade could play a role in the observed differences in IDV seroprevalence among these areas. Indeed, the overall level of exposure in cattle and swine in destination countries (e.g. Italy) is higher than in origin countries (e.g. France), leading to the hypothesis of a viral shedding following the transportation of young cattle abroad and thus contributing to larger diffusion at countries of destination. IDV large geographic circulation in cattle from Northern to more Southern European countries also supports the hypothesis of a viral spread through livestock trade. This review summarizes available data on IDV seroprevalence in Europe collected so far and integrates unpublished data from IDV European surveillance framework of the last decade. In addition, the possible role of livestock trade and biosecurity measures in this pathogen's spread is discussed.


Subject(s)
Cattle Diseases/epidemiology , Goat Diseases/epidemiology , Orthomyxoviridae Infections/veterinary , Sheep Diseases/epidemiology , Swine Diseases/epidemiology , Thogotovirus/physiology , Animals , Cattle , Cattle Diseases/virology , Europe/epidemiology , Goat Diseases/virology , Goats , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Prevalence , Seroepidemiologic Studies , Sheep , Sheep Diseases/virology , Sheep, Domestic , Sus scrofa , Swine , Swine Diseases/virology
10.
Emerg Infect Dis ; 26(1): 104-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31855143

ABSTRACT

We conducted an in-depth characterization of the Nipah virus (NiV) isolate previously obtained from a Pteropus lylei bat in Cambodia in 2003 (CSUR381). We performed full-genome sequencing and phylogenetic analyses and confirmed CSUR381 is part of the NiV-Malaysia genotype. In vitro studies revealed similar cell permissiveness and replication of CSUR381 (compared with 2 other NiV isolates) in both bat and human cell lines. Sequence alignments indicated conservation of the ephrin-B2 and ephrin-B3 receptor binding sites, the glycosylation site on the G attachment protein, as well as the editing site in phosphoprotein, suggesting production of nonstructural proteins V and W, known to counteract the host innate immunity. In the hamster animal model, CSUR381 induced lethal infections. Altogether, these data suggest that the Cambodia bat-derived NiV isolate has high pathogenic potential and, thus, provide insight for further studies and better risk assessment for future NiV outbreaks in Southeast Asia.


Subject(s)
Chiroptera/virology , Henipavirus Infections/veterinary , Nipah Virus/pathogenicity , Animals , Cambodia , Genome, Viral/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , Humans , Nipah Virus/genetics , Phylogeny , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Whole Genome Sequencing
11.
Viruses ; 11(6)2019 06 05.
Article in English | MEDLINE | ID: mdl-31195597

ABSTRACT

Influenza D virus (IDV) has first been identified in 2011 in the USA and was shown to mainly circulate in cattle. While IDV is associated with mild respiratory signs, its prevalence is still unknown. In the present study we show that IDV has been circulating throughout France in cattle and small ruminants, with 47.2% and 1.5% seropositivity, respectively. The high prevalence and moderate pathogenicity of IDV in cattle suggest that it may play an initiating role in the bovine respiratory disease complex.


Subject(s)
Cattle Diseases/virology , Orthomyxoviridae Infections/veterinary , Thogotovirus/immunology , Animals , Antibodies, Viral/immunology , Cattle , Cattle Diseases/epidemiology , France , Orthomyxoviridae Infections/epidemiology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/veterinary , Respiratory Tract Infections/virology , Ruminants , Seroepidemiologic Studies
12.
Biomacromolecules ; 9(7): 1945-50, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18494520

ABSTRACT

The photoluminescence features of Eu(3+)-, Tb(3+)-, Tb(3+)/Eu(3+)-alginate aerogel (hydrogel and alcogel) and Eu(3+)-alginate xerogel hybrids were investigated. The Eu(3+)-alginate aerogel and alcogel exhibit the highest (5)D0 quantum efficiencies (9.9 and 8.2%, respectively), while the hydrogel and xerogel have lower values (5.2 and 5.6%, respectively). The Tb(3+)/Eu(3+) hybrids are multiwavelength emitters in which the emission color can be tuned across the chromaticity diagram from the red toward the yellowish-green spectral regions, crossing the white area by selecting the excitation wavelength.


Subject(s)
Hydrogels/chemistry , Lanthanoid Series Elements/chemistry , Luminescent Agents/chemistry , Alginates , Color , Europium , Gels , Glucuronic Acid , Hexuronic Acids , Light , Luminescent Measurements , Terbium
SELECTION OF CITATIONS
SEARCH DETAIL
...